Next Article in Journal
Unified Classification of Bacterial Colonies on Different Agar Media Based on Hyperspectral Imaging and Machine Learning
Next Article in Special Issue
Identification of Selected Antibiotic Resistance Genes in Two Different Wastewater Treatment Plant Systems in Poland: A Preliminary Study
Previous Article in Journal
Wound Healing Activity of the Essential Oil of Bursera morelensis, in Mice
Previous Article in Special Issue
A Survey of the Presence of Pharmaceutical Residues in Wastewaters. Evaluation of Their Removal using Conventional and Natural Treatment Procedures
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Selected Pharmaceuticals in Different Aquatic Compartments: Part II—Toxicity and Environmental Risk Assessment

LAQV, REQUIMTE, Laboratory of Bromatology and Pharmacognosy, Faculty of Pharmacy, University of Coimbra, Polo III, Azinhaga de St Comba, 3000-548 Coimbra, Portugal
*
Author to whom correspondence should be addressed.
Molecules 2020, 25(8), 1796; https://doi.org/10.3390/molecules25081796
Submission received: 12 March 2020 / Revised: 3 April 2020 / Accepted: 8 April 2020 / Published: 14 April 2020
(This article belongs to the Special Issue Pharmaceutical Residues in the Environment)

Abstract

:
Potential risks associated with releases of human pharmaceuticals into the environment have become an increasingly important issue in environmental health. This concern has been driven by the widespread detection of pharmaceuticals in all aquatic compartments. Therefore, 22 pharmaceuticals, 6 metabolites and transformation products, belonging to 7 therapeutic groups, were selected to perform a review on their toxicity and environmental risk assessment (ERA) in different aquatic compartments, important issues to tackle the water framework directive (WFD). The toxicity data collected reported, with the exception of anxiolytics, at least one toxicity value for concentrations below 1 µg L−1. The results obtained for the ERA revealed risk quotients (RQs) higher than 1 in all the aquatic bodies and for the three trophic levels, algae, invertebrates and fish, posing ecotoxicological pressure in all of these compartments. The therapeutic groups with higher RQs were hormones, antiepileptics, anti-inflammatories and antibiotics. Unsurprisingly, RQs values were highest in wastewaters, however, less contaminated water bodies such as groundwaters still presented maximum values up to 91,150 regarding 17α-ethinylestradiol in fish. Overall, these results present an important input for setting prioritizing measures and sustainable strategies, minimizing their impact in the aquatic environment.

1. Introduction

The environmental impact of medicinal products has been recognized worldwide, and as its use cannot be avoided, a sound risk assessment of their presence in the environment is a key issue that must be tackled to meet the European Union (EU) Water Framework Directive (WFD) [1]. The potential of human pharmaceuticals for negative ecotoxicological effects, even at sublethal concentrations, in the aquatic environment has been of concern since the issue was first brought to attention in 1985 [2]. Nonetheless, the ecotoxicological risks associated to the ubiquitous occurrence of pharmaceuticals in aquatic ecosystems are far from being fully known [3].
According to the European Medicines Agency (EMA) legislation, and since 2006, before a pharmaceutical obtains a marketing authorization approval, it must be demonstrated that it poses no risk to the environment through an environmental risk assessment (ERA). ERA compares the predicted environmental concentrations (PECs), with the predicted no effect concentrations (PNECs) of three trophic levels of aquatic organisms [4,5]. When the pharmaceutical is already on the market, instead of using the PEC, which predict the environmental concentration, we can use the measured environmental concentrations (MEC) reflecting the real concentration in the aquatic environment [6]. Therefore, for marketed pharmaceuticals, high-quality monitoring data, along with data on ecotoxicological and toxicological effects are crucial to perform the ERA, which associates the presence of pharmaceuticals with their impact on the aquatic ecosystem and human health, supporting the selection of possible new priority substances to be monitored [7,8,9].
Thus, a systematic review, in order to provide a clear insight on pharmaceuticals toxicology and on ERA, should embrace, not only several parent compounds, but also, metabolites and transformations products belonging to different therapeutic groups such as: the anxiolytics and hypnotics, further referred only as anxiolytics, alprazolam (ALP), lorazepam (LOR) and zolpidem (ZOL); the antibiotics azithromycin (AZI), ciprofloxacin (CIP), clarithromycin (CLA) and erythromycin (ERY); the lipid regulators bezafibrate (BEZ), gemfibrozil (GEM) and simvastatin (SIM); the antiepileptic carbamazepine (CAR); the selective serotonin reuptake inhibitors (SSRIs) citalopram (CIT) and its main metabolite desmethylcitalopram (N-CIT), escitalopram (ESC), fluoxetine (FLU) and its main metabolite norfluoxetine (Nor-FLU), paroxetine (PAR), sertraline (SER) and its main metabolite desmethylsertraline (Nor-SER); the anti-inflammatories and/or analgesics and antipyretics, further referred only as anti-inflammatories, diclofenac (DIC) and its main metabolite 4-hydroxydiclofenac (4-OH-DIC), ibuprofen (IBU), naproxen (NAP), paracetamol (PARA) and its transformation product 4-aminophenol (4-PARA); and the hormones 17β-estradiol (E2) and its main metabolite estrone (E1) and 17α-ethinylestradiol (EE2; Table 1).
The pharmaceuticals in study, key representatives of major classes of pharmaceuticals, were selected based on the EU watch list, their high consumption, pharmacokinetics, physicochemical properties, persistence, previous studies on the occurrence on the aquatic environment, and their potential toxicological impact, both on humans and on the aquatic environment [10,11,12,13,14]. This review will provide a more realistic water quality assessment contributing for a more integrative approach to rank and prioritize pharmaceuticals, based on an integrated assessment of ERA and exposure in the aquatic environment.
“Water is not a commercial product like any other but, rather, a heritage which must be protected, defended and treated as such”, the claim by the EU WFD contrasts with the poor ecological status of European freshwater bodies, where only 43% achieve a good ecological status. In addition, and despite the enormous efforts, the picture that emerges regarding ecological status is still incomplete, fragmented and with contradictory assessments of the situation. Therefore, it is important to obtain a better understanding of the regional and global context, concerning the environmental risk posed by pharmaceuticals in the aquatic environment.

2. Toxicity

Since pharmaceuticals are continuously introduced into the aquatic environment, they can promote toxic effects on living organisms, even when present at concentrations on the ng L−1 level [15]. This potential for negative effects of pharmaceuticals even at sublethal concentrations, namely for aquatic organisms, has been of concern since the issue was first brought to attention in 1985 [2]. Therefore, their presence poses a threat to the quality of water resources [5,16].
Pharmaceuticals have a relatively clear mode of action in target organisms, and given that fish and invertebrates share more drug targets with humans, it would be expected that they would also respond to pharmaceuticals in a similar way. However, when non-target-species are exposed, unknown effects and potential risks need to be assessed. One example is the impact of EE2 in the feminization of fish [17,18,19]. Nonetheless, all the ecotoxicological risks associated to the ubiquitous occurrence of pharmaceuticals in aquatic ecosystems are far from known [3].
Sorption to sediments is one factor that influences toxicity of pharmaceuticals, although higher sorption to sediments results in an apparent reduction of bioavailability and toxicity, the activity of benthic invertebrate in sediments results in a higher exposure for these organisms [20].
Moreover, bioaccumulation and biomagnification should also be accounted for since they can increase toxicity [17]. These parameters are also related to log Dow, since compounds with values higher than 3 have a tendency for bioaccumulation [17,21], which means that the ionization state can influence the toxicity of pharmaceuticals, and that the pH variability in surface water should also be taken into account [17].
A bibliographic search of the scientific literature was conducted on Google Scholar using the search terms “ecotoxicology” and each of the selected compounds. All the publications that presented ecotoxicological studies on the selected compounds, referring to the concentrations, were included. Below, the ecotoxicological data in the aquatic biota was reviewed, presenting the toxicity data obtained from 120 exposure studies of three trophic levels of non-target organisms, algae (Figure 1), invertebrates (Figure 2) and fish (Figure 3), Table S1 (Supporting information). The data was divided by the different endpoints found in the literature: no observed effect concentrations (NOEC), lowest observed effect concentrations (LOEC), effective concentration (EC50) and lethal concentration (LC50). These endpoints are expected to have increasing concentrations, since they were organized from the more susceptible endpoint (NOEC) to the less one (LC50). However, each endpoint encloses various species of the same trophic level and different toxicological tests like immobilization, growth, luminescence, reproduction, morphology, behavior, etc. When no experimental data was available, L(E)C50 values were estimated with ECOSAR 2.0. This program estimates data on acute toxicity through the molecule structure, sometimes underestimating toxic effects. The data was also divided in acute and chronic toxicity, depending on the time of exposure and trophic level. For algae, acute toxicity was considered when the toxicity tests lasted until 4 days (96 h), longer exposures were considered chronic toxicity. Regarding invertebrates, with the exception of Brachionus calyciflorus (were 2 days was considered chronic data, since it has a shorter life cycle), acute toxicity was accounted when the exposure took place until 2 days (48 h) and chronic toxicity when it was equal or longer than 7 days. For fish, tests until 4 days (96 h) were included in acute toxicity data and exposures equal or above 7 days entered the chronic toxicity data. These criteria were based on OECD tests for each trophic level [22].
Although, as expected, some therapeutic groups presented higher toxicity, such as hormones, which can promote endocrine modifications, all therapeutic groups presented toxicity at low concentrations, highlighting the ecotoxicity of the selected pharmaceuticals [138]. Overall, considering all trophic levels, all therapeutic groups with the exception of anxiolytics, had at least one toxicity report for concentrations below 1 µg L−1, near the concentrations found in the aquatic environment.
Considering the toxicity of the selected pharmaceuticals in all trophic levels, we could observe that the most sensitive one, with the lowest concentrations promoting toxic effects was fish, followed by invertebrates and algae. The limitation of this analysis is that, regarding fish, there were also toxicity data obtained through cell line or tissue testing, which can be difficult to extrapolate to the entire organism. The therapeutic group with higher toxicity, mainly chronic toxicity in fish and invertebrates, are hormones. Additionally, the pharmaceutical that presented higher toxicity, with the lowest concentration promoting toxic effects, was EE2 at 0.1 ng L−1 in fish (NOEC, chronic toxicity) [123]. The highest concentrations promoting toxicity were detected in fish (LC50, acute toxicity), for CLA, CIP and ERY (1 g L−1), [23,123,126].
Ecotoxicological chronic studies on pharmaceuticals are lacking, meaning that many questions about the threat to the environment of pharmaceuticals remain unanswered. Additionally, the actual exposure scenario regards multiple pharmaceuticals, posing uncertainty regarding toxicology in long-term exposure. If many pharmaceuticals are present and share the same mode of action, then the toxicity of this mixture could be higher than if only one pharmaceutical is present, being usually considered the concept of concentration addition, although antagonistic and synergistic effects may also occur. This could result in risk underestimation, as the typical exposure is toward multicomponent chemicals [139,140,141,142].
One example of mixture effects was observed when using a mixture of anti-inflammatories (DIC, IBU and NAP). In this case, the acute toxicity was detected at concentrations where little or no effect was observed for the chemicals individually [20]. Even in mixtures with pharmaceuticals belonging to different therapeutic groups, additive and synergistic effects were reported. A mixture with E2 and FLU promoted a decrease in the reproductive success of D. magna more significantly than either chemical compounds alone [143]. Another example was provided by exposing D. magna to a mixture of CAR and a lipid lowering agent, which exhibited stronger effects during immobilization tests than the single compounds at the same concentration [20].
Taking into account mixture effects, some research has already been developed focusing on toxic effects, and not on specific pharmaceuticals. This was already used to evaluate wastewater treatment plants (WWTPs) removal efficiencies, by evaluating and comparing the toxicity (androgenecity, cytotoxicity, anti-estrogenicity and L. variegatus decrease in reproduction and biomass) both in wastewater influents (WWIs) and efluents (WWEs) [144,145].
Additionally to the active compounds of pharmaceuticals, excipients and additives are also present in medicines, that may contain endocrine disrupting chemical excipients and additives [138].
The measured concentrations of some of the selected pharmaceuticals reported for surface water all over the world surpassed the concentrations here described for toxicity, which suggests that the aquatic biota could be vulnerable to the presence of pharmaceuticals in their environment, and that toxic effects are expected to occur with unexpected outcomes [146].
It is unlikely that pharmaceuticals present in drinking water may pose a risk to the human health through chronic exposure, however, the toxicological implications are not clear [147]. Furthermore, studies have shown that infants may have difficulty in metabolizing drugs therefore, being more vulnerable to the toxic effects of these compounds [17].
As referred, many pharmaceuticals have the potential for bioaccumulation and biomagnification, and chronic effects on ecosystems cannot be ignored for animals at the higher end of the food web [148]. Thus, the health hazard of human exposure by ingestion of contaminated foods should also be taken into account [17].

2.1. Anxiolytics

No ecotoxicological data was found in literature for ALP, LOR and ZOL, and for that reason, all the results for this therapeutic group were obtained from ECOSAR 2.0 [18]. In decreasing order, the more toxic was ZOL, followed by ALP and LOR. The trophic level with the lowest reported concentrations producing toxicity was algae (from 0.211 to 6.07 mg L−1), followed by fish (from 0.248 to 43.1 mg L−1) and invertebrates (from 1.55 to 39.4 mg L−1).

2.2. Antibiotics

Observing the acute toxicity for antibiotics, since there is little data on chronic endpoints, the pattern for the three trophic levels was similar for all antibiotics, with algae being more susceptible at lower concentrations (from 0.0018 to 20.6 mg L−1), followed by invertebrates (from 0.22 to 120 mg L−1) and fish (from 84 to 1000 mg L−1). If we compare each antibiotic, concerning invertebrates, it can be observed that CLA and CIP presented similar results, but when compared with ERY, lower concentrations (220 µg L−1) of this antibiotic can produce the same toxic effects, in this case growth inhibition [23].
In this therapeutic class, in addition to direct toxicological risks, concern has been raised about the potential for the antibiotic residues in water, since they are typically found in the aquatic environment at subtherapeutic concentrations, promoting the emergence of resistant bacteria and subsequent development of more resistant and virulent pathogens [149]. These bacterial resistances, through horizontal gene transfer, may end up in human pathogens, raising questions on human health and the stability of the ecosystem [150,151,152,153,154].
This emergence of bacterial resistance presents one of the major emerging threats to human health and is by far the highest risk for humans of having medicinal products residues in the environment [155]. Furthermore, historical evidence appears to indicate that in the aquatic environment resistance might be acquired faster than in the terrestrial environment [156].
Corroborating the effects on bacteria, changes in biomass and growth rate were reported at concentrations above 5.7 µg L−1 [47]. This therapeutic class can also induce immunotoxicity in the freshwater mussel at low concentrations, between 2 ng L−1 and 1100 ng L−1 [157].

2.3. Lipid Regulators

In this group, the pattern observed with both previous therapeutic groups was not so clear, with median concentrations similar in all trophic levels for acute toxicity. Observing these data, SIM was clearly the pharmaceutical, which promoted toxicity at lower concentrations for invertebrates (160 ng L−1) and fish (765 µg L−1) [26,56]. However, data on chronic toxicity, only available for GEM in two trophic levels, showed that the highest toxicity regarded fish (1.5 µg L−1), followed by invertebrates (78.0 µg L−1) [51,85].

2.4. Antiepileptics

For CAR, once again, the pattern of acute data, was similar to that registered for anxiolytics and antibiotics, with the lowest concentrations promoting toxicity at 10.0 µg L−1, 20 000 µg L−1 and 0.01 µg L−1 for algae, fish and invertebrates, respectively [27,44,57]. Considering the chronic data, similar concentrations were found to produce toxicity in invertebrates and fish trophic levels, ranging from 25 to 25,000 µg L−1 [27,29].

2.5. SSRIs

This therapeutic group has the peculiarity that the phylogenetically ancient and highly conserved neurotransmitter and neurohormone serotonin has been found in invertebrates and vertebrates, although its specific physiological role and mode of action is unknown for many species [158]. Many biological functions within invertebrates, such as reproduction, metabolism, molting and behavior, are under the control of serotonin [159]. Therefore, the pharmaceuticals in this therapeutic group could have tremendous effects on these and other organisms [160]. These facts are in agreement with those found in acute toxicity data found, since for all trophic levels this group had globally the lowest concentrations, which promoted toxic effects, being some of these on reproduction, survival and behavior [161].
When observing these data, the most sensitive trophic level was the invertebrates (0.1 µg L−1), followed by algae (12.1 µg L−1) and fish (72.0 µg L−1) [33,57,92]. In invertebrates, the pharmaceuticals with higher toxicity were FLU (100 ng L−1) and its metabolite Nor-FLU (300 µg L−1) and SER (100 µg L−1). On the other side, PAR was the one with lower toxicity [33,36,57,69]. In algae, the pharmaceutical with highest toxicity was SER, however, in invertebrates, FLU surpassed SER toxicity.
The only metabolite referred in the literature concerning toxicity studies was Nor-FLU, with data for algae and invertebrates. When comparing with FLU (algae and invertebrates), it is clear that the median concentrations inducing toxicity were always lower [162].
Studies performed on SER and FLU demonstrated the influence of pH on toxicity, since the uncharged drug can pass easier through the membrane and act inside the cells, showing a tenfold increased toxicity when shifting the pH closest to their pKa, increasing the nonionized form, from 6.5 to 8.5 and from 7.8 to 9, respectively [17,89,92].

2.6. Anti-Inflammatories

Most anti-inflammatories induce the nonspecific inhibition of prostaglandins. This, in turn, means that there is the potential for effects on any of the normal physiological functions mediated by prostaglandins. In fish, for instance, prostaglandins influence mechanisms of behavior and reproduction and, therefore, they can act as endocrine disruptors or modulators, because they can exert their effects by mimicking or antagonizing the effects of hormones, alter their pattern of synthesis and metabolism and modify hormone receptor levels, leading to possible adverse effects [7,163,164,165]. However, different and unexpected toxicity effects were also observed. One of the first was reported in Pakistan, where a catastrophic decline in the Oriental White-backed Vulture population (95%) originated from the exposure to DIC contaminated live-stock carcasses, which promoted fatal renal disease [98,138].
Overall, excepting anxiolytics, anti-inflammatories were less toxic than the other therapeutic groups. Regarding the lowest concentrations that produced acute toxicity in the three trophic levels, invertebrates had the lowest value (10 ng L−1), followed by algae (10 µg L−1) and fish (90 µg L−1), however, when using median values, the differences become less clear [37,44,57,96]. As for chronic data, higher toxicity was observed in fish (500 ng L−1) and invertebrates (200 µg L−1), when compared with algae (4.01 mg L−1), which is in line with the already referred anti-inflammatories mode of action [70,95].
Data for each anti-inflammatory showed no clear pattern, nonetheless, except for invertebrates, NAP and PARA seemed to have lower toxicity than DIC and IBU. When performing a comparison between DIC and its metabolite (4-OH-DIC) in invertebrates and fish, one could observe that they have similar toxicities. Conversely, PARA transformation product (4-PARA) presented higher toxicity than the parent molecule in all three trophic levels.

2.7. Hormones

Although hormones like E1, E2 and EE2 are mainly used for contraception purposes, the physiological effects are not restricted to effects on reproductive and sexual development, and can target mitochondrial function, energy metabolism and cell cycle control [165].
For acute toxicity, there is only data on algae and invertebrates, and algae presented higher toxicity since the lowest concentration promoting toxic effects was at 162 µg L−1, lower than the 1500 µg L−1 observed in invertebrates [43,78]. Nonetheless, the toxicity promoted by this therapeutic group is mainly expected to be detected through chronic toxicity, however, these data could only be obtained for invertebrates and fish. Considering chronic data, in these two trophic levels, hormones presented higher toxicity than the other therapeutic groups, since the lowest concentrations reported were of 100 ng L−1 and 0.1 ng L−1, for invertebrates and fish, respectively [81,82,123]. It should also be noted that, the highest concentration found that promoted toxicity for fish was also very low (1188 ng L−1) [102].
Individually, there were no differences observed between E1 and E2 toxicity, while EE2 seems the most toxic compound regarding chronic toxicity in invertebrates and especially in fish, where the 36 results available presented concentrations below 44 ng L−1 [135]. Namely, when two different fish species were exposed to EE2 at 3 ng L−1 and 4 ng L−1 they suffered sex gender reversal, from male to female, which can strongly unbalance the aquatic ecosystem [130,136,138].

3. Environmental Risk Assessment

The data regarding occurrence and toxicity already presented is crucial in order to perform the ERA, and can be used to select the pharmaceuticals that are more prone to induce toxic effects in aquatic biota [166]. The risk assessment, mentioned in the EMA guideline on the ERA of medicinal products for human use [4], is performed through the risk quotient (RQ) calculation, dividing the PEC by the PNEC for each pharmaceutical, observing three different trophic levels (algae, invertebrates and fish). If RQ is equal or above 1, there is a potential environmental risk situation, whereas when values are lower than 1, no risk is expected. However, a certain risk could be expected for the substances with a RQ between 0.1 and 1 [167,168]. However, this guideline is only applied for marketing authorizations and for pharmaceuticals marketed after 2006. Additionally, it does not constitute a valid criterion upon which to base the refusal of a market authorization of medicinal products for human use in the EU [6]. Our evaluation of the potential ecotoxicological risk posed for the aquatic compartment was based on a dual approach: one using the worst case scenario, as stated by the EMA guideline on the ERA [4], where the maximum individual concentrations of pharmaceuticals found in the respective aquatic compartment were used as MEC [15,169,170], and another using the median concentrations for each pharmaceutical as MEC [171]. This evaluation can also be an important tool to suggest the inclusion or removal of pharmaceuticals in the watch list of the Directive 2013/39/EU.
As discussed, some concentrations compiled in surface water are higher than the levels that induce toxicity, not applying any uncertainty factor (UF) for the PNEC calculation. Additionally, some studies have indicated that concentrations of several pharmaceuticals belonging to different therapeutic groups can promote toxic effects on negatively impacted aquatic biota, presenting RQ higher than 1 [11,18,139,157,172,173].
As referred, aquatic biota inhabiting the receiving environment are unintentionally exposed throughout their lifetime to a complex mixture of residual pharmaceuticals and these mixtures can exhibit a greater effect than individual compounds [20,174,175]. Therefore, it is a challenge to address the concerns related to the chronic effect, low-level exposure to these compounds, including exposure of sensitive subpopulations to pharmaceutical mixtures [17,174].

3.1. Predicted No-Effect Concentration (PNECs)

Based on the toxicity data (Figure 4, Figure 5, Figure 6 and Figure 7), Table 2 presents the PNECs for the selected pharmaceuticals. These values were calculated by applying an UF of 100 and 10 to the long-term EC50 and NOEC values, and an UF of 50 and 1000 to the short-term LOEC and L(E)C50 values, respectively, available in the literature. The UF is an expression of the degree of uncertainty in the extrapolation from the test data on a limited number of species to the actual environment [4]. As referred, when no experimental data are available, L(E)C50 values were estimated through ECOSAR 2.0.
It should be taken into account that the choice of toxicity data can obviously affect the outcome [15]. However, the results obtained for the PNECs were directly related to the toxicity data, and a similar pattern to the toxicity data was observed, with the therapeutic groups and pharmaceuticals with higher toxicity presenting the lowest PNEC values.

3.2. Risk Assessment

Using the occurrence data obtained from Part I of this review (Table S2, Supporting information) and the PNECs previously calculated (Table 2), RQs were deemed for all the selected pharmaceuticals in the different aquatic compartments and are presented in Figure 4, Figure 5, Figure 6 and Figure 7 [146].
In general, the results revealed that RQs higher than 1 could be observed for all the aquatic bodies, posing ecotoxicological pressure in all of these compartments.

3.2.1. Wastewater Influents

The RQs observed in WWIs were the highest from all the aquatic compartments, as well as the concentrations of the selected pharmaceuticals (Figure 4). The highest value (274,816) and median (13,400) were observed for IBU in invertebrates. Anti-inflammatories were the therapeutic group with the highest RQs median, both for the maximum and median values, followed by antiepileptic and hormones.
With the exception of anxiolytics, all the other therapeutic groups presented RQs > 1 for at least two trophic levels, being SSRIs and anxiolytics the only groups that did not present risk RQs > 1 for all pharmaceuticals.
For the anxiolytics and antibiotics, the algae were clearly the most susceptible trophic level, presenting higher RQs. Another clear pattern was observed for the hormones, where for fish all the RQs (median and maximum) were higher than 1 and with the maximum values between 129 and 17 271
These results demonstrated that the concentrations reaching the WWTPs could clearly endanger all the trophic levels that might be exposed to this aquatic matrix.

3.2.2. Wastewater Effluents

This aquatic compartment presents lower RQs than the WWIs (Figure 5). Antibiotics along with antiepileptic, anti-inflammatories and hormones were the therapeutic groups with highest RQs values. When considering median values alone, antibiotics have lower RQs than the other three therapeutic groups. The values observed for antibiotics can promote an even bigger problem than the direct toxicity to aquatic organism: the emergence of bacterial resistance. Nonetheless, the ERA approach does not address this issue [155].
Anxiolytics continued to present RQs values lower than 1. Antibiotics, on the other hand, had the highest value observed for CIP in algae (100,258). As in WWI, antiepileptic continued to present the highest median RQs.
In WWI, some metabolites of the SSRIs presented similar or slightly higher RQs than the parent compounds (N-CIT and Nor-SER), in WWE a similar pattern was observed. These results highlight the fact that parent compounds and metabolites reached WWEs, and that the concentrations found in this matrix were able to promote toxic effects in the aquatic biota. This fact suggests that metabolites and transformation products should also be monitored in the environment.
Regarding the anti-inflammatories therapeutic group, DIC and IBU stand out from the other pharmaceuticals presenting clearly higher median and maximum RQs. As for hormones and observing the fish trophic level, as in the WWI, all RQs were higher than one.

3.2.3. Surface Waters

This aquatic body was clearly more problematic to the environment when compared to WWIs and WWEs since here is where most of aquatic life inhabits. However, like in the previous water compartments, RQs higher than 1 were observed for all trophic levels and therapeutic groups, with the exception of the anxiolytics. Antiepileptic, anti-inflammatories, antibiotics and hormones remained the therapeutic groups with the highest RQs (Figure 6).
The highest maximum and median values regarded IBU (16,327) and CAR (138) in invertebrates. There were still eighteen pharmaceuticals (AZI, CLA, CIP, ERY, BEZ, GEM, SIM, CAR, CIT, FLU, SER, DIC, IBU, PARA, E1, E2, αE2 and EE2) with RQs above 1.
Antibiotics still presented all of their maximum RQs higher than 1 for algae, whereas lipid regulators presented the same pattern for invertebrates. As for the antiepileptic, their maximum values were above one for algae and invertebrates. In SSRIs therapeutic group, CIT, FLU and SER were the ones that presented RQs higher than 1, contributing to the possible risk posed by this group. The hormones, regarding fish, still presented all median RQs higher than 1, with the exception of E1, with the EE2 obtaining the highest RQs for this trophic level.
As already mentioned, this water body, encompassing rivers and lakes, should be free from risk. Nonetheless, from the 28 detected pharmaceuticals, 18 presented maximum RQs above 1 and even 8 had median RQs superior to 1, posing a threat to all the aquatic organisms.

3.2.4. Other Water Bodies

The results obtained for the RQs of other water bodies are presented in Figure 7. The RQs, in decreasing order, were groundwater, seawater, mineral water and drinking water. Since drinking water is usually obtained through surface and groundwaters, these results suggest that the sources used to produce drinking water were the ones with lower pharmaceutical contaminations or that water treatment plants were removing the selected pharmaceuticals. The results obtained in seawater can be biased, since fresh water organisms were used to evaluate the risk and there are reports that marine organism can be more vulnerable, increasing the risk in this water matrix [178,179].
All of these compartments presented values above 1 for all trophic levels, with the exception of mineral water in algae, highlighting, once again, the pressure sustained by the aquatic organisms in all the aquatic compartments. This indicates that there is also a possible risk for humans. The therapeutic groups with higher maximum and median RQs are hormones, antiepileptic, antibiotics and anti-inflammatories, being the same groups as in WWIs.
With the exception of anxiolytics, all therapeutic groups still present RQ medians above 1, with 12 pharmaceuticals with maximum values above 1, and 11 with medians also higher than 1. If we use the threshold of 0.1, where some risk might be expected, we can find 16 of these compounds. Some RQs are still extremely high, with EE2 presenting values up to 91,150 and a median of 6091 for fish in groundwater. The higher RQs observed in these compartments were for CAR and IBU in invertebrates, and for hormones in fish.
Viewing these results, it is possible to observe not only high risk for aquatic organism in wastewaters but, despite the RQ reduction, several pharmaceuticals still promote risk in other supposedly cleaner aquatic matrices. Additionally, additive or even synergistic effects can occur, especially in the pharmaceuticals with the same mechanism of action [139,140,141,142].

3.3. Mitigation Measures

The RQs obtained for all the water compartments, particularly the RQs higher than 1 in surface waters and in the other water bodies, raise not only the issue of toxicity for the aquatic environment but also for humans using these aquatic bodies as a source of drinking water and also for whom eating animals living in these contaminated environments. Therefore, mitigation measures should be implemented to prevent high RQs in these important water resources. These measures should begin through the awareness of the problem. For example, in Sweden, an environmental classification system for drugs has been established through collaboration between producers, authorities and the public health care. This system assesses the environmental risk of pharmaceuticals being publicly available, therefore, the market can demand for medicines with less environmental impact, stimulating producers to design more environmentally friendly medicines [17]. This includes the concept of green pharmacy, where the design of pharmaceutical products focus also on their high metabolization and environmental degradation, reducing the environmental burden and improving environmental safety and health impacts [138].
Additionally, the ERA guideline on human pharmaceuticals should be revised in order to: enter the risk benefit analysis; impose its revision every five years with the new available data; incorporate the pharmaceuticals marketed before 2006 and include metabolites and transformation products [6].
Another issue already referred is the possible improvement of WWTPs removal efficiencies and the improvement of toxicity studies. The main challenges regarding the assessment of ecotoxicity are the scarce information available for some of the selected pharmaceuticals, namely chronic data and effects on multiple life stages or even multiple generations, which mimics the environmental exposure. Behavior studies are also lacking, before death and other major toxicity effects there can be diminished sexual interest, fear or activity, increased curiosity, etc. Although this seems like small behavior changes, it can be enough to unbalance an ecosystem eliminating one or more species by being unable to escape from predators or by lack of reproduction. This leads to another point that ecotoxicity studies should also be performed in ecosystems, because some effects previously referred are more evident in this type of studies. It should also be considered the increase of species tested and mixture effects, not only between different pharmaceuticals of the same therapeutic group but also from different groups and with other substances, like heavy metals. Given the pharmaceutical environmental presence in mixtures and with other substances, additive or even synergistic effects occur; therefore, the real hazard may be greater than that calculated [24,33,180,181]. Another issue that is neglected with the traditional ecotoxicity studies is the emergence of bacterial resistance. We are testing antibiotics in algae, invertebrates and fish but probably the biggest impact will be on bacteria, namely on the emergence of resistant bacteria that can reach not only aquatic animals but also humans. Additionally, the emergence of bacterial resistance is a major concern involving the presence of pharmaceuticals in the aquatic compartment, which is more prone in this environment [6,182].
This could generate benefits in water resource management, by providing the means for cross-compliance measures in environmental regulation and providing an adequate risk assessment for pharmaceuticals mixtures [183]. In this way, the complete scenario of the contamination of pharmaceuticals in the aquatic environment and their risk could be performed, contributing to future improvements in minimization measures and legislation.

4. Final Remarks

A literature review was conducted in order to understand the toxicity and ERA of pharmaceuticals in the aquatic environment. In this context, a broad and specialized background was obtained, enabling an overview of the state of the art in these subjects.
Regarding the toxicity data, although the differences observed between different therapeutic groups and within each therapeutic group, all therapeutic groups with the exception of anxiolytics, had at least one toxicity report for concentrations below 1 µg L−1. The trophic level with the lowest concentrations promoting toxic effects was fish, followed by invertebrates and algae, emphasizing that fish, the trophic level closer to humans, are more prone to toxicity effects from the selected pharmaceuticals.
The results also show that pharmaceuticals with higher RQs are not the ones with higher occurrence and that proper toxicity data is important to a correct evaluation of the ERA.
The ERA performed for the pharmaceuticals in the different aquatic compartments revealed, with the exception of anxiolytics, RQs higher than 1, not only for WWIs but also for all aquatic compartments, for all trophic levels and therapeutic groups.
The therapeutic groups with the highest RQs in all aquatic compartments are hormones, antiepileptics, anti-inflammatories and antibiotics and the pharmaceuticals with the highest values are the EE2, CAR, IBU, CIP and AZI in all aquatic compartments. Highlighting threat to all the aquatic organisms exposed, namely on the feminization of fish by EE2 and its impact on aquatic ecosystems. Additionally, two antibiotics are among the pharmaceuticals with higher RQs and the ERA does not evaluate the emergence of bacterial resistance. If this issue was also evaluated it would probably confirm why they are considered the therapeutic group with the highest risk for humans, regarding the residues of medicinal products in the environment.
Unfortunately, the pressure of pharmaceuticals on aquatic bodies will continue to rise, and, therefore, mitigation measures and changes in legislation must be implemented.

Supplementary Materials

The following are available online: Table S1: Ecotoxicological data on the selected pharmaceuticals, Table S2: Occurrence data from the different aquatic compartments for environmental risk assessment evaluation.

Author Contributions

Conceptualization, A.P. (André Pereira), L.S. and A.P. (Angelina Pena); Methodology, A.P. (André Pereira), L.S. and A.P. (Angelina Pena); Formal analysis, A.P. (André Pereira); Writing—original draft preparation, A.P. (André Pereira); Writing—review and editing, A.P. (André Pereira), L.S. and C.L. (Celia Laranjeiro); Supervision, C.L. (Celeste Lino) and A.P. (Angelina Pena); Project administration, A.P. (Angelina Pena). All authors have read and agreed to the published version of the manuscript.

Funding

The work was supported by UIDB/50006/2020 with funding from FCT/MCTES through national funds.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Robles-Molina, J.; Lara-Ortega, F.J.; Gilbert-López, B.; García-Reyes, J.F.; Molina-Díaz, A. Multi-residue method for the determination of over 400 priority and emerging pollutants in water and wastewater by solid-phase extraction and liquid chromatography-time-of-flight mass spectrometry. J. Chromatogr. A 2014, 1350, 30–43. [Google Scholar] [CrossRef] [PubMed]
  2. Silva, L.J.G.; Lino, C.M.; Meisel, L.M.; Pena, A. Selective serotonin re-uptake inhibitors (SSRIs) in the aquatic environment: An ecopharmacovigilance approach. Sci. Total Environ. 2012, 437, 185–195. [Google Scholar] [CrossRef] [PubMed]
  3. Gonzalez-Rey, M.; Bebianno, M.J. Does selective serotonin reuptake inhibitor (SSRI) fluoxetine affects mussel Mytilus galloprovincialis? Environ. Pollut. 2013, 173, 200–209. [Google Scholar] [CrossRef]
  4. European Medicines Agency Guideline on the Environmental Risk Assessment of Medicinal Products for Human Use; European Medicines Agency: London, UK, 2006.
  5. Taylor, D.; Senac, T. Human pharmaceutical products in the environment—The “problem” in perspective. Chemosphere 2014, 115, 95–99. [Google Scholar] [CrossRef]
  6. Pereira, A.M.P.T.; Silva, L.J.G.; Lino, C.M.; Meisel, L.M.; Pena, A. A critical evaluation of different parameters for estimating pharmaceutical exposure seeking an improved environmental risk assessment. Sci. Total Environ. 2017, 603–604, 226–236. [Google Scholar] [CrossRef]
  7. Mompelat, S.; Le Bot, B.; Thomas, O. Occurrence and fate of pharmaceutical products and by-products, from resource to drinking water. Environ. Int. 2009, 35, 803–814. [Google Scholar] [CrossRef]
  8. Van Doorslaer, X.; Dewulf, J.; Van Langenhove, H.; Demeestere, K. Fluoroquinolone antibiotics: An emerging class of environmental micropollutants. Sci. Total Environ. 2014, 500–501, 250–269. [Google Scholar] [CrossRef]
  9. Boxall, A.B.A.; Keller, V.D.J.; Straub, J.O.; Monteiro, S.C.; Fussell, R.; Williams, R.J. Exploiting monitoring data in environmental exposure modelling and risk assessment of pharmaceuticals. Environ. Int. 2014, 73, 176–185. [Google Scholar] [CrossRef] [Green Version]
  10. European Commission Implementing Decision (EU) 2018/840 of 5 June 2018 establishing a watch list of substances for Union-wide monitoring in the field of water policy pursuant to Directive 2008/105/EC of the European Parliament and of the Council and repealing 495. Off. J. Eur. Union 2018, L141, 9–12.
  11. Papageorgiou, M.; Kosma, C.; Lambropoulou, D. Seasonal occurrence, removal, mass loading and environmental risk assessment of 55 pharmaceuticals and personal care products in a municipal wastewater treatment plant in Central Greece. Sci. Total Environ. 2016, 543, 547–569. [Google Scholar] [CrossRef]
  12. Ter Laak, T.L.; Van der Aa, M.; Houtman, C.J.; Stoks, P.G.; Van Wezel, A.P. Relating environmental concentrations of pharmaceuticals to consumption: A mass balance approach for the river Rhine. Environ. Int. 2010, 36, 403–409. [Google Scholar] [CrossRef] [PubMed]
  13. Bade, R.; Rousis, N.I.; Bijlsma, L.; Gracia-Lor, E.; Castiglioni, S.; Sancho, J.V.; Hernandez, F. Screening of pharmaceuticals and illicit drugs in wastewater and surface waters of Spain and Italy by high resolution mass spectrometry using UHPLC-QTOF MS and LC-LTQ-Orbitrap MS. Anal. Bioanal. Chem. 2015, 407, 8979–8988. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Ribeiro, A.R.; Nunes, O.C.; Pereira, M.F.R.; Silva, A.M.T. An overview on the advanced oxidation processes applied for the treatment of water pollutants defined in the recently launched Directive 2013/39/EU. Environ. Int. 2015, 75, 33–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Pereira, A.M.P.T.; Silva, L.J.G.; Meisel, L.M.; Lino, C.M.; Pena, A. Environmental impact of pharmaceuticals from Portuguese wastewaters: Geographical and seasonal occurrence, removal and risk assessment. Environ. Res. 2015, 136, 108–119. [Google Scholar] [CrossRef] [Green Version]
  16. He, Y.; Sutton, N.B.; Rijnaarts, H.H.H.; Langenhoff, A.A.M. Degradation of pharmaceuticals in wastewater using immobilized TiO2 photocatalysis under simulated solar irradiation. Appl. Catal. B Environ. 2016, 182, 132–141. [Google Scholar] [CrossRef]
  17. Zenker, A.; Cicero, M.R.; Prestinaci, F.; Bottoni, P.; Carere, M. Bioaccumulation and biomagnification potential of pharmaceuticals with a focus to the aquatic environment. J. Environ. Manag. 2014, 133, 378–387. [Google Scholar] [CrossRef]
  18. Vergeynst, L.; Haeck, A.; De Wispelaere, P.; Van Langenhove, H.; Demeestere, K. Multi-residue analysis of pharmaceuticals in wastewater by liquid chromatography-magnetic sector mass spectrometry: Method quality assessment and application in a Belgian case study. Chemosphere 2015, 119, S2–S8. [Google Scholar] [CrossRef]
  19. Johnson, A.C.; Sumpter, J.P. Putting pharmaceuticals into the wider context of challenges to fish populations in rivers. Philos. Trans. R. Soc. B Biol. Sci. 2014, 369, 20130581. [Google Scholar] [CrossRef] [Green Version]
  20. Petrie, B.; Barden, R.; Kasprzyk-Hordern, B. A review on emerging contaminants in wastewaters and the environment: Current knowledge, understudied areas and recommendations for future monitoring. Water Res. 2014, 72, 3–27. [Google Scholar] [CrossRef]
  21. Lavén, M.; Alsberg, T.; Yu, Y.; Adolfsson-Erici, M.; Sun, H. Serial mixed-mode cation- and anion-exchange solid-phase extraction for separation of basic, neutral and acidic pharmaceuticals in wastewater and analysis by high-performance liquid chromatography-quadrupole time-of-flight mass spectrometry. J. Chromatogr. A 2009, 1216, 49–62. [Google Scholar] [CrossRef]
  22. OECD Guidance Document on the Use of the Harmonised System for the Classification of Chemicals Which Are Hazardous for the Aquatic Environment; OECD: Paris, France, 2001.
  23. Isidori, M.; Lavorgna, M.; Nardelli, A.; Pascarella, L.; Parrella, A. Toxic and genotoxic evaluation of six antibiotics on non-target organisms. Sci. Total Environ. 2005, 346, 87–98. [Google Scholar] [CrossRef] [PubMed]
  24. Yang, L.-H.; Ying, G.-G.; Su, H.-C.; Stauber, J.L.; Adams, M.S.; Binet, M.T. Growth-inhibiting effects of 12 antibacterial agenst and their mixtures on the freshwater microalga Pseudokirchneriella subcapitata. Environ. Toxicol. Chem. 2008, 27, 1201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Farré, M.; Ferrer, I.; Ginebreda, A.; Figueras, M.; Olivella, L.; Tirapu, L.; Vilanova, M.; Barceló, D. Determination of drugs in surface water and wastewater samples by liquid chromatography–mass spectrometry: Methods and preliminary results including toxicity studies with Vibrio fischeri. J. Chromatogr. A 2001, 938, 187–197. [Google Scholar] [CrossRef]
  26. DeLorenzo, M.E.; Fleming, J. Individual and mixture effects of selected pharmaceuticals and personal care products on the marine phytoplankton species Dunaliella tertiolecta. Arch. Environ. Contam. Toxicol. 2008, 54, 203–210. [Google Scholar] [CrossRef] [PubMed]
  27. Ferrari, B.; Mons, R.; Vollat, B.; Fraysse, B.; Paxéus, N.; Lo Giudice, R.; Pollio, A.; Garric, J. Environmental risk assessment of six human pharmaceuticals: Are the current environmental risk assessment procedures sufficient for the protection of the aquatic environment? Environ. Toxicol. Chem. 2004, 23, 1344–1354. [Google Scholar] [CrossRef] [Green Version]
  28. Cleuvers, M. Aquatic ecotoxicity of pharmaceuticals including the assessment of combination effects. Toxicol. Lett. 2003, 142, 185–194. [Google Scholar] [CrossRef]
  29. Ferrari, B.; Paxéus, N.; Giudice, R.L.; Pollio, A.; Garric, J. Ecotoxicological impact of pharmaceuticals found in treated wastewaters: Study of carbamazepine, clofibric acid, and diclofenac. Ecotoxicol. Environ. Saf. 2003, 55, 359–370. [Google Scholar] [CrossRef]
  30. Christensen, A.M.; Faaborg-Andersen, S.; Ingerslev, F.; Baun, A. Mixture and single-substance toxicity of selective serotonin reuptake inhibitors toward algae and crustaceans. Environ. Toxicol. Chem. 2007, 26, 85. [Google Scholar] [CrossRef] [Green Version]
  31. Brooks, B.W.; Turner, P.K.; Stanley, J.K.; Weston, J.J.; Glidewell, E.A.; Foran, C.M.; Slattery, M.; La Point, T.W.; Huggett, D.B. Waterborne and sediment toxicity of fluoxetine to select organisms. Chemosphere 2003, 52, 135–142. [Google Scholar] [CrossRef]
  32. Brooks, B.W.; Foran, C.M.; Richards, S.M.; Weston, J.; Turner, P.K.; Stanley, J.K.; Solomon, K.R.; Slattery, M.; La Point, T.W. Aquatic ecotoxicology of fluoxetine. Toxicol. Lett. 2003, 142, 169–183. [Google Scholar] [CrossRef]
  33. Johnson, D.J.; Sanderson, H.; Brain, R.A.; Wilson, C.J.; Solomon, K.R. Toxicity and hazard of selective serotonin reuptake inhibitor antidepressants fluoxetine, fluvoxamine, and sertraline to algae. Ecotoxicol. Environ. Saf. 2007, 67, 128–139. [Google Scholar] [CrossRef]
  34. Neuwoehner, J.; Fenner, K.; Escher, B.I. Physiological modes of action of fluoxetine and its human metabolites in algae. Environ. Sci. Technol. 2009, 43, 6830–6837. [Google Scholar] [CrossRef]
  35. Halling-Sorensen, B. Environmental risk assessment of antibiotics: Comparison of mecillinam, trimethoprim and ciprofloxacin. J. Antimicrob. Chemother. 2000, 46, 53–58. [Google Scholar] [CrossRef] [Green Version]
  36. Minagh, E.; Hernan, R.; O’Rourke, K.; Lyng, F.M.; Davoren, M. Aquatic ecotoxicity of the selective serotonin reuptake inhibitor sertraline hydrochloride in a battery of freshwater test species. Ecotoxicol. Environ. Saf. 2009, 72, 434–440. [Google Scholar] [CrossRef] [PubMed]
  37. Lawrence, J.R.; Swerhone, G.D.W.; Topp, E.; Korber, D.R.; Neu, T.R.; Wassenaar, L.I. Structural and functional responses of river biofilm communities to the nonsteroidal anti-inflammatory diflofenac. Environ. Toxicol. Chem. 2007, 26, 573. [Google Scholar] [CrossRef]
  38. Ortiz de García, S.; Pinto, G.P.; García-Encina, P.A.; Mata, R.I. Ranking of concern, based on environmental indexes, for pharmaceutical and personal care products: An application to the Spanish case. J. Environ. Manage. 2013, 129, 384–397. [Google Scholar] [CrossRef] [PubMed]
  39. Pomati, F.; Netting, A.G.; Calamari, D.; Neilan, B.A. Effects of erythromycin, tetracycline and ibuprofen on the growth of Synechocystis sp. and Lemna minor. Aquat. Toxicol. 2004, 67, 387–396. [Google Scholar] [CrossRef] [PubMed]
  40. Isidori, M.; Lavorgna, M.; Nardelli, A.; Parrella, A.; Previtera, L.; Rubino, M. Ecotoxicity of naproxen and its phototransformation products. Sci. Total Environ. 2005, 348, 93–101. [Google Scholar] [CrossRef]
  41. Cleuvers, M. Mixture toxicity of the anti-inflammatory drugs diclofenac, ibuprofen, naproxen, and acetylsalicylic acid. Ecotoxicol. Environ. Saf. 2004, 59, 309–315. [Google Scholar] [CrossRef]
  42. Henschel, K.-P.; Wenzel, A.; Diedrich, M.; Fliedner, A. Environmental Hazard Assessment of Pharmaceuticals. Regul. Toxicol. Pharmacol. 1997, 25, 220–225. [Google Scholar] [CrossRef] [PubMed]
  43. Onogbosele, C.O. Bioavailability of organic contaminants in rivers. Ph.D. Thesis, Brunel University, London, UK, 2015. [Google Scholar]
  44. Li, Y.; Zhang, L.; Liu, X.; Ding, J. Ranking and prioritizing pharmaceuticals in the aquatic environment of China. Sci. Total Environ. 2019, 658, 333–342. [Google Scholar] [CrossRef] [PubMed]
  45. Machado, M.D.; Soares, E.V. Sensitivity of freshwater and marine green algae to three compounds of emerging concern. J. Appl. Phycol. 2019, 31, 399–408. [Google Scholar] [CrossRef] [Green Version]
  46. Martins, N.; Pereira, R.; Abrantes, N.; Pereira, J.; Gonçalves, F.; Marques, C.R. Ecotoxicological effects of ciprofloxacin on freshwater species: Data integration and derivation of toxicity thresholds for risk assessment. Ecotoxicology 2012, 21, 1167–1176. [Google Scholar] [CrossRef] [PubMed]
  47. Ebert, I.; Bachmann, J.; Kühnen, U.; Küster, A.; Kussatz, C.; Maletzki, D.; Schlüter, C. Toxicity of the fluoroquinolone antibiotics enrofloxacin and ciprofloxacin to photoautotrophic aquatic organisms. Environ. Toxicol. Chem. 2011, 30, 2786–2792. [Google Scholar] [CrossRef]
  48. Brain, R.A.; Johnson, D.J.; Richards, S.M.; Sanderson, H.; Sibley, P.K.; Solomon, K.R. Effects of 25 pharmaceutical compounds to Lemna gibba using a seven-day static-renewel test. Environ. Toxicol. Chem. 2004, 23, 371–382. [Google Scholar] [CrossRef]
  49. Robinson, A.A.; Belden, J.B.; Lydy, M.J. Toxicity of fluoroquinolone antibiotics to aquatic organisms. Environ. Toxicol. Chem. 2005, 24, 423. [Google Scholar] [CrossRef]
  50. Nie, X.; Wang, X.; Chen, J.; Zitko, V.; An, T. Response of the freshwater Alga chlorella vulgaris to trichloroisocyanuric acid and ciprofloxacin. Environ. Toxicol. Chem. 2008, 27, 168–173. [Google Scholar] [CrossRef]
  51. Isidori, M.; Nardelli, A.; Pascarella, L.; Rubino, M.; Parrella, A. Toxic and genotoxic impact of fibrates and their photoproducts on non-target organisms. Environ. Int. 2007, 33, 635–641. [Google Scholar] [CrossRef]
  52. Zurita, J.L.; Repetto, G.; Jos, A.; Salguero, M.; López-Artíguez, M.; Cameán, A.M. Toxicological effects of the lipid regulator gemfibrozil in four aquatic systems. Aquat. Toxicol. 2007, 81, 106–115. [Google Scholar] [CrossRef]
  53. Quinn, B.; Gagné, F.; Blaise, C. An investigation into the acute and chronic toxicity of eleven pharmaceuticals (and their solvents) found in wastewater effluent on the cnidarian, Hydra attenuata. Sci. Total Environ. 2008, 389, 306–314. [Google Scholar] [CrossRef]
  54. Han, G.H.; Hur, H.G.; Kim, S.D. Ecotoxicological risk of pharmaceuticals from wastewater treatment plants in Korea: Occurrence and toxicity to Daphnia magna. Environ. Toxicol. Chem. 2006, 25, 265–271. [Google Scholar] [CrossRef]
  55. Key, P.B.; Hoguet, J.; Reed, L.A.; Chung, K.W.; Fulton, M.H. Effects of the statin antihyperlipidemic agent simvastatin on grass shrimp, Palaemonetes pugio. Environ. Toxicol. 2008, 23, 153–160. [Google Scholar] [CrossRef]
  56. Dahl, U.; Gorokhova, E.; Breitholtz, M. Application of growth-related sublethal endpoints in ecotoxicological assessments using a harpacticoid copepod. Aquat. Toxicol. 2006, 77, 433–438. [Google Scholar] [CrossRef] [PubMed]
  57. De Lange, H.J.; Noordoven, W.; Murk, A.J.; Lürling, M.; Peeters, E.T.H.M. Behavioural responses of Gammarus pulex (Crustacea, Amphipoda) to low concentrations of pharmaceuticals. Aquat. Toxicol. 2006, 78, 209–216. [Google Scholar] [CrossRef] [PubMed]
  58. Kim, J.-W.; Ishibashi, H.; Yamauchi, R.; Ichikawa, N.; Takao, Y.; Hirano, M.; Koga, M.; Arizono, K. Acute toxicity of pharmaceutical and personal care products on freshwater crustacean (Thamnocephalus platyurus) and fish (Oryzias latipes). J. Toxicol. Sci. 2009, 34, 227–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Kim, Y.; Choi, K.; Jung, J.; Park, S.; Kim, P.-G.; Park, J. Aquatic toxicity of acetaminophen, carbamazepine, cimetidine, diltiazem and six major sulfonamides, and their potential ecological risks in Korea. Environ. Int. 2007, 33, 370–375. [Google Scholar] [CrossRef]
  60. Fong, P.P.; Molnar, N. Antidepressants cause foot detachment from substrate in five species of marine snail. Mar. Environ. Res. 2013, 84, 24–30. [Google Scholar] [CrossRef]
  61. Nałecz-Jawecki, G. Evaluation of the in vitro biotransformation of fluoxetine with HPLC, mass spectrometry and ecotoxicological tests. Chemosphere 2007, 70, 29–35. [Google Scholar] [CrossRef]
  62. Nentwig, G. Effects of pharmaceuticals on aquatic invertebrates. Part II: The antidepressant drug fluoxetine. Arch. Environ. Contam. Toxicol. 2007, 52, 163–170. [Google Scholar] [CrossRef]
  63. Péry, A.R.R.; Gust, M.; Vollat, B.; Mons, R.; Ramil, M.; Fink, G.; Ternes, T.; Garric, J. Fluoxetine effects assessment on the life cycle of aquatic invertebrates. Chemosphere 2008, 73, 300–304. [Google Scholar] [CrossRef] [Green Version]
  64. Gust, M.; Buronfosse, T.; Giamberini, L.; Ramil, M.; Mons, R.; Garric, J. Effects of fluoxetine on the reproduction of two prosobranch mollusks: Potamopyrgus antipodarum and Valvata piscinalis. Environ. Pollut. 2009, 157, 423–429. [Google Scholar] [CrossRef]
  65. Hazelton, P.D.; Cope, W.G.; Mosher, S.; Pandolfo, T.J.; Belden, J.B.; Barnhart, M.C.; Bringolf, R.B. Fluoxetine alters adult freshwater mussel behavior and larval metamorphosis. Sci. Total Environ. 2013, 445–446, 94–100. [Google Scholar] [CrossRef]
  66. Cunningham, V.L.; Constable, D.J.C.; Hannah, R.E. Environmental Risk Assessment of Paroxetine. Environ. Sci. Technol. 2004, 38, 3351–3359. [Google Scholar] [CrossRef]
  67. Lamichhane, K.; Garcia, S.N.; Huggett, D.B.; DeAngelis, D.L.; La Point, T.W. Exposures to a selective serotonin reuptake inhibitor (SSRI), sertraline hydrochloride, over multiple generations: Changes in life history traits in Ceriodaphnia dubia. Ecotoxicol. Environ. Saf. 2014, 101, 124–130. [Google Scholar] [CrossRef]
  68. Cleuvers, M. Chronic Mixture Toxicity of Pharmaceuticals to Daphnia—The Example of Nonsteroidal Anti-Inflammatory Drugs. In Pharmaceuticals in the Environment; Kümmerer, K., Ed.; Springer: Berlin, Heidelberg, 2006; pp. 277–284. ISBN 978-3-540-74664-5. [Google Scholar]
  69. Nalecz-Jawecki, G.; Persoone, G. Toxicity of selected pharmaceuticals to the Anostracan crustacean Thamnocephalus platyurus—Comparison of sublethal and lethal effect levels with the 1 h Rapidtoxkit and the 24 h Thamnotoxkit microbiotests. Environ. Sci. Pollut. Res. Int. 2006, 13, 22–27. [Google Scholar] [CrossRef]
  70. Haap, T.; Triebskorn, R.; Köhler, H.-R. Acute effects of diclofenac and DMSO to Daphnia magna: Immobilisation and hsp70-induction. Chemosphere 2008, 73, 353–359. [Google Scholar] [CrossRef]
  71. Heckmann, L.-H.; Callaghan, A.; Hooper, H.L.; Connon, R.; Hutchinson, T.H.; Maund, S.J.; Sibly, R.M. Chronic toxicity of ibuprofen to Daphnia magna: Effects on life history traits and population dynamics. Toxicol. Lett. 2007, 172, 137–145. [Google Scholar] [CrossRef]
  72. Pounds, N.; Maclean, S.; Webley, M.; Pascoe, D.; Hutchinson, T. Acute and chronic effects of ibuprofen in the mollusc Planorbis carinatus (Gastropoda: Planorbidae). Ecotoxicol. Environ. Saf. 2008, 70, 47–52. [Google Scholar] [CrossRef]
  73. Dave, G.; Herger, G. Determination of detoxification to Daphnia magna of four pharmaceuticals and seven surfactants by activated sludge. Chemosphere 2012, 88, 459–466. [Google Scholar] [CrossRef]
  74. Sherer, J.T. Pharmaceuticals in the environment. Am. J. Heal. Pharm. 2006, 63, 174–178. [Google Scholar] [CrossRef]
  75. El-Bassat, R.; Touliabah, H.; Harisa, G. Toxicity of four pharmaceuticals from different classes to isolated plankton species. African J. Aquat. Sci. 2012, 37, 71–80. [Google Scholar] [CrossRef]
  76. Li, M.-H. Acute toxicity of 30 pharmaceutically active compounds to freshwater planarians, Dugesia japonica. Toxicol. Environ. Chem. 2013, 95, 1157–1170. [Google Scholar] [CrossRef]
  77. Kühn, R.; Pattard, M.; Pernak, K.; Winter, A. Results of the harmful effects of selected water pollutants (anilines, phenols, aliphatic compounds) to Daphnia magna. Water Res. 1989, 23, 495–499. [Google Scholar] [CrossRef]
  78. Li, M. Acute toxicity of industrial endocrine-disrupting chemicals, natural and synthetic sex hormones to the freshwater planarian, Dugesia japonica. Toxicol. Environ. Chem. 2013, 95, 984–991. [Google Scholar] [CrossRef]
  79. Jaser, W. Effects of 17α-ethinylestradiol on the reproduction of the cladoceran species Ceriodaphnia reticulata and Sida crystallina. Environ. Int. 2003, 28, 633–638. [Google Scholar] [CrossRef]
  80. Caldwell, D.J.; Mastrocco, F.; Hutchinson, T.H.; Länge, R.; Heijerick, D.; Janssen, C.; Anderson, P.D.; Sumpter, J.P. Derivation of an Aquatic Predicted No-Effect Concentration for the Synthetic Hormone, 17α-Ethinyl Estradiol. Environ. Sci. Technol. 2008, 42, 7046–7054. [Google Scholar] [CrossRef] [Green Version]
  81. Vandenbergh, G.F.; Adriaens, D.; Verslycke, T.; Janssen, C.R. Effects of 17α-ethinylestradiol on sexual development of the amphipod Hyalella azteca. Ecotoxicol. Environ. Saf. 2003, 54, 216–222. [Google Scholar] [CrossRef]
  82. Jobling, S.; Casey, D.; Rodgers-Gray, T.; Oehlmann, J.; Schulte-Oehlmann, U.; Pawlowski, S.; Baunbeck, T.; Turner, A.; Tyler, C. Comparative responses of molluscs and fish to environmental estrogens and an estrogenic effluent. Aquat. Toxicol. 2004, 66, 207–222. [Google Scholar] [CrossRef]
  83. Sidhu, H.; O’Connor, G.; McAvoy, D. Risk assessment of biosolids-borne ciprofloxacin and azithromycin. Sci. Total Environ. 2019, 651, 3151–3160. [Google Scholar] [CrossRef]
  84. Henry, T.B.; Kwon, J.W.; Armbrust, K.L.; Black, M.C. Acute and chronic toxicity of five selective serotonin reuptake inhibitors in Ceriodaphnia dubia. Environ. Toxicol. Chem. 2004, 23, 2229–2233. [Google Scholar] [CrossRef]
  85. Mimeault, C.; Trudeau, V.L.; Moon, T.W. Waterborne gemfibrozil challenges the hepatic antioxidant defense system and down-regulates peroxisome proliferator-activated receptor beta (PPARbeta) mRNA levels in male goldfish (Carassius auratus). Toxicology 2006, 228, 140–150. [Google Scholar] [CrossRef]
  86. Raldúa, D.; André, M.; Babin, P.J. Clofibrate and gemfibrozil induce an embryonic malabsorption syndrome in zebrafish. Toxicol. Appl. Pharmacol. 2008, 228, 301–314. [Google Scholar]
  87. Caminada, D.; Escher, C.; Fent, K. Cytotoxicity of pharmaceuticals found in aquatic systems: Comparison of PLHC-1 and RTG-2 fish cell lines. Aquat. Toxicol. 2006, 79, 114–123. [Google Scholar] [CrossRef]
  88. Richards, S.M.; Cole, S.E. A toxicity and hazard assessment of fourteen pharmaceuticals to Xenopus laevis larvae. Ecotoxicology 2006, 15, 647–656. [Google Scholar] [CrossRef]
  89. Stanley, J.K.; Ramirez, A.J.; Chambliss, C.K.; Brooks, B.W. Enantiospecific sublethal effects of the antidepressant fluoxetine to a model aquatic vertebrate and invertebrate. Chemosphere 2007, 69, 9–16. [Google Scholar] [CrossRef]
  90. Henry, T.B.; Black, M.C. Acute and chronic toxicity of fluoxetine (selective serotonin reuptake inhibitor) in western mosquitofish. Arch. Environ. Contam. Toxicol. 2008, 54, 325–330. [Google Scholar] [CrossRef]
  91. Nakamura, Y.; Yamamoto, H.; Sekizawa, J.; Kondo, T.; Hirai, N.; Tatarazako, N. The effects of pH on fluoxetine in Japanese medaka (Oryzias latipes): Acute toxicity in fish larvae and bioaccumulation in juvenile fish. Chemosphere 2008, 70, 865–873. [Google Scholar] [CrossRef]
  92. Valenti, T.W.; Hurtado, P.P.; Chambliss, C.K.; Brooks, B.W. Aquatic toxicity of sertraline to Pimephales promelas at environmentally relevant surface water pH. Environ. Toxicol. Chem. 2009, 28, 2685–2694. [Google Scholar] [CrossRef]
  93. Schwaiger, J.; Ferling, H.; Mallow, U.; Wintermayr, H.; Negele, R.D. Toxic effects of the non-steroidal anti-inflammatory drug diclofenac. Part I: Histopathological alterations and bioaccumulation in rainbow trout. Aquat. Toxicol. 2004, 68, 141–150. [Google Scholar] [CrossRef]
  94. Triebskorn, R.; Casper, H.; Heyd, A.; Eikemper, R.; Köhler, H.-R.; Schwaiger, J. Toxic effects of the non-steroidal anti-inflammatory drug diclofenac. Part II: Cytological effects in liver, kidney, gills and intestine of rainbow trout (Oncorhynchus mykiss). Aquat. Toxicol. 2004, 68, 151–166. [Google Scholar] [CrossRef]
  95. Hoeger, B.; Köllner, B.; Dietrich, D.R.; Hitzfeld, B. Water-borne diclofenac affects kidney and gill integrity and selected immune parameters in brown trout (Salmo trutta f. fario). Aquat. Toxicol. 2005, 75, 53–64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Memmert, U.; Peither, A.; Burri, R.; Weber, K.; Schmidt, T.; Sumpter, J.P.; Hartmann, A. Diclofenac: New data on chronic toxicity and bioconcentration in fish. Environ. Toxicol. Chem. 2013, 32, 442–452. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Mehinto, A.C.; Hill, E.M.; Tyler, C.R. Uptake and biological effects of environmentally relevant concentrations of the nonsteroidal anti-inflammatory pharmaceutical diclofenac in rainbow trout (Oncorhynchus mykiss). Environ. Sci. Technol. 2010, 44, 2176–2182. [Google Scholar] [CrossRef]
  98. Oaks, J.L.; Gilbert, M.; Virani, M.Z.; Watson, R.T.; Meteyer, C.U.; Rideout, B.A.; Shivaprasad, H.L.; Ahmed, S.; Iqbal Chaudhry, M.J.; Arshad, M.; et al. Diclofenac residues as the cause of vulture population decline in Pakistan. Nature 2004, 427, 630–633. [Google Scholar] [CrossRef] [PubMed]
  99. Li, Q.; Wang, P.; Chen, L.; Gao, H.; Wu, L. Acute toxicity and histopathological effects of naproxen in zebrafish (Danio rerio) early life stages. Environ. Sci. Pollut. Res. 2016, 23, 18832–18841. [Google Scholar] [CrossRef]
  100. Sun, L.W.; Qu, M.M.; Li, Y.Q.; Wu, Y.L.; Chen, Y.G.; Kong, Z.M.; Liu, Z.T. Toxic effects of aminophenols on aquatic life using the Zebrafish embryo test and the comet assay. Bull. Environ. Contam. Toxicol. 2004, 73, 628–634. [Google Scholar] [CrossRef] [PubMed]
  101. Metcalfe, C.D.; Metcalfe, T.L.; Kiparissis, Y.; Koenig, B.G.; Khan, C.; Hughes, R.J.; Croley, T.R.; March, R.E.; Potter, T. Estrogenic potency of chemicals detected in sewage treatment plant effluents as determined by in vivo assays with Japanese medaka ( Oryzias latipes ). Environ. Toxicol. Chem. 2001, 20, 297–308. [Google Scholar] [CrossRef]
  102. Imai, S.; Koyama, J.; Fujii, K. Effects of estrone on full life cycle of Java medaka (Oryzias javanicus), a new marine test fish. Environ. Toxicol. Chem. 2007, 26, 726–731. [Google Scholar] [CrossRef]
  103. Thorpe, K.L.; Benstead, R.; Hutchinson, T.H.; Tyler, C.R. Associations between altered vitellogenin concentrations and adverse health effects in fathead minnow (Pimephales promelas). Aquat. Toxicol. 2007, 85, 176–183. [Google Scholar] [CrossRef]
  104. Kang, I.J.; Yokota, H.; Oshima, Y.; Tsuruda, Y.; Yamaguchi, T.; Maeda, M.; Imada, N.; Tadokoro, H.; Honjo, T. Effect of 17β-estradiol on the reproduction of Japanese medaka (Oryzias latipes). Chemosphere 2002, 47, 71–80. [Google Scholar] [CrossRef]
  105. Caldwell, D.J.; Mastrocco, F.; Anderson, P.D.; Länge, R.; Sumpter, J.P. Predicted-no-effect concentrations for the steroid estrogens estrone, 17β-estradiol, estriol, and 17α-ethinylestradiol. Environ. Toxicol. Chem. 2012, 31, 1396–1406. [Google Scholar] [CrossRef] [PubMed]
  106. Kramer, V.J.; Miles-Richardson, S.; Pierens, S.L.; Giesy, J.P. Reproductive impairment and induction of alkaline-labile phosphate, a biomarker of estrogen exposure, in fathead minnows (Pimephales promelas) exposed to waterborne 17β-estradiol. Aquat. Toxicol. 1998, 40, 335–360. [Google Scholar] [CrossRef]
  107. Shappell, N.W.; Elder, K.H.; West, M. Estrogenicity and nutrient concentration of surface waters surrounding a large confinement dairy operation using best management practices for land application of animal wastes. Environ. Sci. Technol. 2010, 44, 2365–2371. [Google Scholar] [CrossRef]
  108. Seki, M.; Fujishima, S.; Nozaka, T.; Maeda, M.; Kobayashi, K. Comparison of response to 17β-estradiol and 17β-trenbolone among three small fish species. Environ. Toxicol. Chem. 2006, 25, 2742. [Google Scholar] [CrossRef] [PubMed]
  109. Brion, F.; Tyler, C.; Palazzi, X.; Laillet, B.; Porcher, J.; Garric, J.; Flammarion, P. Impacts of 17β-estradiol, including environmentally relevant concentrations, on reproduction after exposure during embryo-larval-, juvenile- and adult-life stages in zebrafish (Danio rerio). Aquat. Toxicol. 2004, 68, 193–217. [Google Scholar] [PubMed]
  110. Van der Ven, L.T.M.; Van den Brandhof, E.-J.; Vos, J.H.; Wester, P.W. Effects of the estrogen agonist 17β-estradiol and antagonist tamoxifen in a partial life-cycle assay with with zebrafish (danio rerio). Environ. Toxicol. Chem. 2007, 26, 92. [Google Scholar] [CrossRef] [PubMed]
  111. Nash, J.P.; Kime, D.E.; Van der Ven, L.T.M.; Wester, P.W.; Brion, F.; Maack, G.; Stahlschmidt-Allner, P.; Tyler, C.R. Long-term exposure to environmental concentrations of the pharmaceutical ethynylestradiol causes reproductive failure in fish. Environ. Health Perspect. 2004, 112, 1725–1733. [Google Scholar] [CrossRef] [Green Version]
  112. Hirai, N.; Nanba, A.; Koshio, M.; Kondo, T.; Morita, M.; Tatarazako, N. Feminization of Japanese medaka (Oryzias latipes) exposed to 17β-estradiol: Formation of testis–ova and sex-transformation during early-ontogeny. Aquat. Toxicol. 2006, 77, 78–86. [Google Scholar] [CrossRef]
  113. Seki, M.; Yokota, H.; Maeda, M.; Kobayashi, K. Fish full life-cycle testing for 17beta-estradiol on medaka (Oryzias latipes). Environ. Toxicol. Chem. 2005, 24, 1259–1266. [Google Scholar] [CrossRef]
  114. Jukosky, J.A.; Watzin, M.C.; Leiter, J.C. The effects of environmentally relevant mixtures of estrogens on Japanese medaka (Oryzias latipes) reproduction. Aquat. Toxicol. 2008, 86, 323–331. [Google Scholar] [CrossRef]
  115. Imai, S.; Koyama, J.; Fujii, K. Effects of 17β-estradiol on the reproduction of Java-medaka (Oryzias javanicus), a new test fish species. Mar. Pollut. Bull. 2005, 51, 708–714. [Google Scholar] [CrossRef] [PubMed]
  116. Cripe, G.M.; Hemmer, B.L.; Goodman, L.R.; Fournie, J.W.; Raimondo, S.; Vennari, J.C.; Danner, R.L.; Smith, K.; Manfredonia, B.R.; Kulaw, D.H.; et al. Multigerational exposure of the estuarine Sheepshead minnow (cyprinodon variegatus) to 17β-estradiol. I. Organism-level effects over three generations. Environ. Toxicol. Chem. 2009, 28, 2397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Toft, G.; Baatrup, E. Altered sexual characteristics in guppies (Poecilia reticulata) exposed to 17β-estradiol and 4-tert-octylphenol during sexual development. Ecotoxicol. Environ. Saf. 2003, 56, 228–237. [Google Scholar] [CrossRef]
  118. Liao, T.; Guo, Q.L.; Jin, S.W.; Cheng, W.; Xu, Y. Comparative responses in rare minnow exposed to 17β-estradiol during different life stages. Fish Physiol. Biochem. 2009, 35, 341–349. [Google Scholar] [CrossRef] [PubMed]
  119. Robinson, C.D.; Brown, E.; Craft, J.A.; Davies, I.M.; Megginson, C.; Miller, C.; Moffat, C.F. Bioindicators and reproductive effects of prolonged 17β-oestradiol exposure in a marine fish, the sand goby (Pomatoschistus minutus). Aquat. Toxicol. 2007, 81, 397–408. [Google Scholar] [CrossRef]
  120. Pollino, C.A.; Georgiades, E.; Holdway, D.A. Use of the Australian crimson-spotted rainbowfish (Melanotaenia fluviatilis) as a model test species for investigating the effects of endocrine disruptors. Environ. Toxicol. Chem. 2007, 26, 2171. [Google Scholar] [CrossRef] [PubMed]
  121. Pawlowski, S.; van Aerle, R.; Tyler, C.; Braunbeck, T. Effects of 17α-ethinylestradiol in a fathead minnow (Pimephales promelas) gonadal recrudescence assay. Ecotoxicol. Environ. Saf. 2004, 57, 330–345. [Google Scholar] [CrossRef]
  122. Örn, S.; Holbech, H.; Madsen, T.H.; Norrgren, L.; Petersen, G.I. Gonad development and vitellogenin production in zebrafish (Danio rerio) exposed to ethinylestradiol and methyltestosterone. Aquat. Toxicol. 2003, 65, 397–411. [Google Scholar] [CrossRef]
  123. Zha, J.; Sun, L.; Zhou, Y.; Spear, P.A.; Ma, M.; Wang, Z. Assessment of 17α-ethinylestradiol effects and underlying mechanisms in a continuous, multigeneration exposure of the Chinese rare minnow (Gobiocypris rarus). Toxicol. Appl. Pharmacol. 2008, 226, 298–308. [Google Scholar] [CrossRef]
  124. Schäfers, C.; Teigeler, M.; Wenzel, A.; Maack, G.; Fenske, M.; Segner, H. Concentration- and time-dependent effects of the synthetic estrogen, 17α-ethinylestradiol, on reproductive capabilities of the Zebrafish, Danio rerio. J. Toxicol. Environ. Heal. Part A 2007, 70, 768–779. [Google Scholar] [CrossRef]
  125. Van den Belt, K.; Berckmans, P.; Vangenechten, C.; Verheyen, R.; Witters, H. Comparative study on the in vitro/in vivo estrogenic potencies of 17β-estradiol, estrone, 17α-ethynylestradiol and nonylphenol. Aquat. Toxicol. 2004, 66, 183–195. [Google Scholar] [CrossRef] [PubMed]
  126. Xu, H.; Yang, J.; Wang, Y.; Jiang, Q.; Chen, H.; Song, H. Exposure to 17α-ethynylestradiol impairs reproductive functions of both male and female zebrafish (Danio rerio). Aquat. Toxicol. 2008, 88, 1–8. [Google Scholar] [CrossRef] [PubMed]
  127. Soares, J.; Coimbra, A.M.; Reis-Henriques, M.A.; Monteiro, N.M.; Vieira, M.N.; Oliveira, J.M.A.; Guedes-Dias, P.; Fontaínhas-Fernandes, A.; Parra, S.S.; Carvalho, A.P. Disruption of zebrafish (Danio rerio) embryonic development after full life-cycle parental exposure to low levels of ethinylestradiol. Aquat. Toxicol. 2009, 95, 330–338. [Google Scholar] [CrossRef]
  128. Parrott, J.L.; Blunt, B.R. Life-cycle exposure of fathead minnows (Pimephales promelas) to an ethinylestradiol concentration below 1 ng/L reduces egg fertilization success and demasculinizes males. Environ. Toxicol. 2005, 20, 131–141. [Google Scholar] [CrossRef]
  129. Länge, R.; Hutchinson, T.H.; Croudace, C.P.; Siegmund, F.; Schweinfurth, H.; Hampe, P.; Panter, G.H.; Sumpter, J.P. Effects of the synthetic estrogen 17 alpha-ethinylestradiol on the life-cycle of the fathead minnow (Pimephales promelas). Environ. Toxicol. Chem. 2001, 20, 1216–1227. [Google Scholar] [CrossRef] [Green Version]
  130. Fenske, M.; Maack, G.; Schäfers, C.; Segner, H. An environmentally relevant concentration of estrogen induces arrest of male gonad development in zebrafish, Danio rerio. Environ. Toxicol. Chem. 2005, 24, 1088–1098. [Google Scholar] [CrossRef]
  131. Scholz, S. 17-α-ethinylestradiol affects reproduction, sexual differentiation and aromatase gene expression of the medaka (Oryzias latipes). Aquat. Toxicol. 2000, 50, 363–373. [Google Scholar] [CrossRef]
  132. Balch, G.C.; Mackenzie, C.A.; Metcalfe, C.D. Alterations of gonadal development and reproductive success in Japanese medaka (Oryzias latipes) exposed to 17α-ethinylestradiol. Environ. Toxicol. Chem. 2004, 23, 782. [Google Scholar] [CrossRef]
  133. Schultz, I.R.; Skillman, A.; Nicolas, J.-M.; Cyr, D.G.; Nagler, J.J. Short-term exposure to 17α-ethynylestradiol decreases the fertility of sexually maturing male rainbow trout ( Oncorhynchus mykiss ). Environ. Toxicol. Chem. 2003, 22, 1272–1280. [Google Scholar] [CrossRef] [PubMed]
  134. Zillioux, E.J.; Johnson, I.C.; Kiparissis, Y.; Metcalfe, C.D.; Wheat, J.V.; Ward, S.G.; Liu, H. The sheepshead minnow as an in vivo model for endocrine disruption in marine teleosts: A partial life-cycle test with 17α-ethynylestradiol. Environ. Toxicol. Chem. 2001, 20, 1968–1978. [Google Scholar] [CrossRef] [PubMed]
  135. Kristensen, T.; Baatrup, E.; Bayley, M. 17α-Ethinylestradiol Reduces the Competitive Reproductive Fitness of the Male Guppy (Poecilia reticulata)1. Biol. Reprod. 2005, 72, 150–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Lange, A.; Katsu, Y.; Ichikawa, R.; Paull, G.C.; Chidgey, L.L.; Coe, T.S.; Iguchi, T.; Tyler, C.R. Altered sexual development in roach (Rutilus rutilus) exposed to environmental concentrations of the pharmaceutical 17α-Ethinylestradiol and associated expression dynamics of aromatases and estrogen receptors. Toxicol. Sci. 2008, 106, 113–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Yang, X.; Xu, X.; Wei, X.; Wan, J.; Zhang, Y. Biomarker effects in carassius auratus exposure to ofloxacin, sulfamethoxazole and ibuprofen. Int. J. Environ. Res. Public Health 2019, 16, 1628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. Kümmerer, K. Pharmaceuticals in the Environment. Annu. Rev. Environ. Resour. 2010, 35, 57–75. [Google Scholar] [CrossRef] [Green Version]
  139. Oliveira, T.S.; Murphy, M.; Mendola, N.; Wong, V.; Carlson, D.; Waring, L. Characterization of Pharmaceuticals and Personal Care products in hospital effluent and waste water influent/effluent by direct-injection LC-MS-MS. Sci. Total Environ. 2015, 518–519, 459–478. [Google Scholar] [CrossRef] [PubMed]
  140. Backhaus, T.; Faust, M. Predictive environmental risk assessment of chemical mixtures: A conceptual framework. Environ. Sci. Technol. 2012, 46, 2564–2573. [Google Scholar] [CrossRef]
  141. Escher, B.I.; Baumgartner, R.; Koller, M.; Treyer, K.; Lienert, J.; McArdell, C.S. Environmental toxicology and risk assessment of pharmaceuticals from hospital wastewater. Water Res. 2011, 45, 75–92. [Google Scholar] [CrossRef] [Green Version]
  142. Jones-Lepp, T.L.; Sanchez, C.; Alvarez, D.A.; Wilson, D.C.; Taniguchi-Fu, R.L. Point sources of emerging contaminants along the Colorado River Basin: Source water for the arid Southwestern United States. Sci. Total Environ. 2012, 430, 237–245. [Google Scholar] [CrossRef] [Green Version]
  143. McEachran, A.D.; Shea, D.; Bodnar, W.; Nichols, E.G. Pharmaceutical occurrence in groundwater and surface waters in forests land-applied with municipal wastewater. Environ. Toxicol. Chem. 2016, 35, 898–905. [Google Scholar] [CrossRef] [Green Version]
  144. Stalter, D.; Magdeburg, A.; Wagner, M.; Oehlmann, J. Ozonation and activated carbon treatment of sewage effluents: Removal of endocrine activity and cytotoxicity. Water Res. 2011, 45, 1015–1024. [Google Scholar] [CrossRef]
  145. Magdeburg, A.; Stalter, D.; Oehlmann, J. Whole effluent toxicity assessment at a wastewater treatment plant upgraded with a full-scale post-ozonation using aquatic key species. Chemosphere 2012, 88, 1008–1014. [Google Scholar] [CrossRef] [PubMed]
  146. Pereira, A.; Silva, L.; Laranjeiro, C.; Lino, C.; Pena, A. Selected Pharmaceuticals in Different Aquatic Compartments: Part I—Source, Fate and Occurrence. Molecules 2020, 25, 1026. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Benotti, M.J.; Trenholm, R.A.; Vanderford, B.J.; Holady, J.C.; Stanford, B.D.; Snyder, S.A. Pharmaceuticals and endocrine disrupting compounds in U.S. drinking water. Environ. Sci. Technol. 2009, 43, 597–603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Crouse, B.A.; Ghoshdastidar, A.J.; Tong, A.Z. The presence of acidic and neutral drugs in treated sewage effluents and receiving waters in the Cornwallis and Annapolis River watersheds and the Mill CoveSewage Treatment Plant in Nova Scotia, Canada. Environ. Res. 2012, 112, 92–99. [Google Scholar] [CrossRef] [PubMed]
  149. Tuševljak, N.; Dutil, L.; Rajić, A.; Uhland, F.C.; McClure, C.; St-Hilaire, S.; Reid-Smith, R.J.; McEwen, S.A. Antimicrobial Use and Resistance in Aquaculture: Findings of a Globally Administered Survey of Aquaculture-Allied Professionals. Zoonoses Public Health 2013, 60, 426–436. [Google Scholar] [CrossRef]
  150. Kostich, M.S.; Batt, A.L.; Lazorchak, J.M. Concentrations of prioritized pharmaceuticals in effluents from 50 large wastewater treatment plants in the US and implications for risk estimation. Environ. Pollut. 2014, 184, 354–359. [Google Scholar] [CrossRef] [PubMed]
  151. Khan, G.A.; Berglund, B.; Khan, K.M.; Lindgren, P.E.; Fick, J. Occurrence and Abundance of Antibiotics and Resistance Genes in Rivers, Canal and near Drug Formulation Facilities—A Study in Pakistan. PLoS ONE 2013, 8, e62712. [Google Scholar] [CrossRef]
  152. Zuccato, E.; Castiglioni, S.; Bagnati, R.; Melis, M.; Fanelli, R. Source, occurrence and fate of antibiotics in the Italian aquatic environment. J. Hazard. Mater. 2010, 179, 1042–1048. [Google Scholar] [CrossRef]
  153. Rizzo, L.; Manaia, C.; Merlin, C.; Schwartz, T.; Dagot, C.; Ploy, M.C.; Michael, I.; Fatta-Kassinos, D. Urban wastewater treatment plants as hotspots for antibiotic resistant bacteria and genes spread into the environment: A review. Sci. Total Environ. 2013, 447, 345–360. [Google Scholar] [CrossRef]
  154. Fick, J.; Söderström, H. Contamination of surface, ground, and drinking water from pharmaceutical production. Environ. Toxicol. Chem. 2009, 28, 2522–2527. [Google Scholar] [CrossRef]
  155. EAHC Study on the Environmental Risks of Medicinal Products Executive Agency for Health and Consumers Document Information; Executive Agency for Health and Consumers: Luxembourg, 2013.
  156. Cabello, F.C. Heavy use of prophylactic antibiotics in aquaculture: A growing problem for human and animal health and for the environment. Environ. Microbiol. 2006, 8, 1137–1144. [Google Scholar] [CrossRef] [PubMed]
  157. Segura, P.A.; Takada, H.; Correa, J.A.; El Saadi, K.; Koike, T.; Onwona-Agyeman, S.; Ofosu-Anim, J.; Sabi, E.B.; Wasonga, O.V.; Mghalu, J.M.; et al. Global occurrence of anti-infectives in contaminated surface waters: Impact of income inequality between countries. Environ. Int. 2015, 80, 89–97. [Google Scholar] [CrossRef]
  158. Kreke, N.; Dietrich, D.R. Physiological endpoints for potential SSRI interactions in fish. Crit. Rev. Toxicol. 2008, 38, 215–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  159. Bossus, M.C.; Guler, Y.Z.; Short, S.J.; Morrison, E.R.; Ford, A.T. Behavioural and transcriptional changes in the amphipod Echinogammarus marinus exposed to two antidepressants, fluoxetine and sertraline. Aquat. Toxicol. 2014, 151, 46–56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  160. Lajeunesse, A.; Gagnon, C.; Sauvé, S. Determination of Basic Antidepressants and Their N-Desmethyl Metabolites in Raw Sewage and Wastewater Using Solid-Phase Extraction and Liquid Chromatography-Tandem Mass Spectrometry. Anal. Chem. 2008, 80, 5325–5333. [Google Scholar] [CrossRef] [PubMed]
  161. Weinberger, J.; Klaper, R. Environmental concentrations of the selective serotonin reuptake inhibitor fluoxetine impact specific behaviors involved in reproduction, feeding and predator avoidance in the fish Pimephales promelas (fathead minnow). Aquat. Toxicol. 2014, 151, 77–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Celiz, M.D.; Tso, J.; Aga, D.S. Pharmaceutical metabolites in the environment: Analytical challenges and ecological risks. Environ. Toxicol. Chem. 2009, 28, 2473. [Google Scholar] [CrossRef]
  163. Laganà, A.; Bacaloni, A.; De Leva, I.; Faberi, A.; Fago, G.; Marino, A. Analytical methodologies for determining the occurrence of endocrine disrupting chemicals in sewage treatment plants and natural waters. Anal. Chim. Acta 2004, 501, 79–88. [Google Scholar] [CrossRef]
  164. Fent, K.; Weston, A.A.; Caminada, D. Ecotoxicology of human pharmaceuticals. Aquat. Toxicol. 2006, 76, 122–159. [Google Scholar] [CrossRef]
  165. Corcoran, J.; Winter, M.J.; Tyler, C.R. Pharmaceuticals in the aquatic environment: A critical review of the evidence for health effects in fish. Crit. Rev. Toxicol. 2010, 40, 287–304. [Google Scholar] [CrossRef]
  166. Pereira, A.M.P.T.; Silva, L.J.G.; Laranjeiro, C.S.M.; Meisel, L.M.; Lino, C.M.; Pena, A. Human pharmaceuticals in Portuguese rivers: The impact of water scarcity in the environmental risk. Sci. Total Environ. 2017, 609, 1182–1191. [Google Scholar] [CrossRef] [PubMed]
  167. Bound, J.P.; Voulvoulis, N. Predicted and measured concentrations for selected pharmaceuticals in UK rivers: Implications for risk assessment. Water Res. 2006, 40, 2885–2892. [Google Scholar] [CrossRef] [PubMed]
  168. Holm, G.; Snape, J.R.; Murray-Smith, R.; Talbot, J.; Taylor, D.; Sörme, P. Implementing ecopharmacovigilance in practice: Challenges and potential opportunities. Drug Saf. 2013, 36, 533–546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  169. Santos, L.H.M.L.M.; Gros, M.; Rodriguez-Mozaz, S.; Delerue-Matos, C.; Pena, A.; Barceló, D.; Montenegro, M.C.B.S.M. Contribution of hospital effluents to the load of pharmaceuticals in urban wastewaters: Identification of ecologically relevant pharmaceuticals. Sci. Total Environ. 2013, 461–462, 302–316. [Google Scholar] [CrossRef] [PubMed]
  170. Vazquez-Roig, P.; Andreu, V.; Blasco, C.; Picó, Y. Risk assessment on the presence of pharmaceuticals in sediments, soils and waters of the Pego-Oliva Marshlands (Valencia, eastern Spain). Sci. Total Environ. 2012, 440, 24–32. [Google Scholar] [CrossRef]
  171. Pereira, A.M.P.T.; Silva, L.J.G.; Lino, C.M.; Meisel, L.M.; Pena, A. Assessing environmental risk of pharmaceuticals in Portugal: An approach for the selection of the Portuguese monitoring stations in line with Directive 2013/39/EU. Chemosphere 2016, 144, 2507–2515. [Google Scholar] [CrossRef]
  172. Zhang, R.; Zhang, G.; Zheng, Q.; Tang, J.; Chen, Y.; Xu, W.; Zou, Y.; Chen, X. Occurrence and risks of antibiotics in the Laizhou Bay, China: Impacts of river discharge. Ecotoxicol. Environ. Saf. 2012, 80, 208–215. [Google Scholar] [CrossRef]
  173. Acuña, V.; Schiller, D.; García-Galán, M.J.; Rodríguez-Mozaz, S.; Corominas, L.; Petrovic, M.; Poch, M.; Barceló, D.; Sabater, S. Occurrence and in-stream attenuation of wastewater-derived pharmaceuticals in Iberian rivers. Sci. Total Environ. 2015, 503–504, 133–141. [Google Scholar] [CrossRef] [Green Version]
  174. Simazaki, D.; Kubota, R.; Suzuki, T.; Akiba, M.; Nishimura, T.; Kunikane, S. Occurrence of selected pharmaceuticals at drinking water purification plants in Japan and implications for human health. Water Res. 2015, 76, 187–200. [Google Scholar] [CrossRef]
  175. Henry, T.B.; Black, M.C. Mixture and single-substance acute toxicity of selective serotonin reuptake inhibitors in Ceriodaphnia dubia. Environ. Toxicol. Chem. 2007, 26, 1751–1755. [Google Scholar] [CrossRef] [Green Version]
  176. Vestel, J.; Caldwell, D.J.; Constantine, L.; D’Aco, V.J.; Davidson, T.; Dolan, D.G.; Millard, S.P.; Murray-Smith, R.; Parke, N.J.; Ryan, J.J.; et al. Use of acute and chronic ecotoxicity data in environmental risk assessment of pharmaceuticals. Environ. Toxicol. Chem. 2016, 35, 1201–1212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Schultz, M.M.; Painter, M.M.; Bartell, S.E.; Logue, A.; Furlong, E.T.; Werner, S.L.; Schoenfuss, H.L. Selective uptake and biological consequences of environmentally relevant antidepressant pharmaceutical exposures on male fathead minnows. Aquat. Toxicol. 2011, 104, 38–47. [Google Scholar] [CrossRef] [PubMed]
  178. Minguez, L.; Pedelucq, J.; Farcy, E.; Ballandonne, C.; Budzinski, H.; Halm-Lemeille, M.P. Toxicities of 48 pharmaceuticals and their freshwater and marine environmental assessment in northwestern France. Environ. Sci. Pollut. Res. 2016, 23, 4992–5001. [Google Scholar] [CrossRef] [PubMed]
  179. Gao, P.; Li, Z.; Gibson, M.; Gao, H. Ecological risk assessment of nonylphenol in coastal waters of China based on species sensitivity distribution model. Chemosphere 2014, 104, 113–119. [Google Scholar] [CrossRef]
  180. Richards, S.M.; Wilson, C.J.; Johnson, D.J.; Castle, D.M.; Lam, M.; Mabury, S.A.; Sibley, P.K.; Solomon, K.R. Effects of pharmaceutical mixtures in aquatic microcosms. Environ. Toxicol. Chem. 2004, 23, 1035–1042. [Google Scholar] [CrossRef] [Green Version]
  181. Goolsby, E.W.; Mason, C.M.; Wojcik, J.T.; Jordan, A.M.; Black, M.C. Acute and chronic effects of diphenhydramine and sertraline mixtures in Ceriodaphnia dubia. Environ. Toxicol. Chem. 2013, 32, 2866–2869. [Google Scholar] [CrossRef]
  182. Pereira, A.M.P.T.; Silva, L.J.G.; Meisel, L.M.; Pena, A. Fluoroquinolones and Tetracycline Antibiotics in a Portuguese Aquaculture System and Aquatic Surroundings: Occurrence and Environmental Impact. J. Toxicol. Environ. Heal. Part A 2015, 78, 959–975. [Google Scholar] [CrossRef]
  183. Altenburger, R.; Ait-Aissa, S.; Antczak, P.; Backhaus, T.; Barceló, D.; Seiler, T.-B.; Brion, F.; Busch, W.; Chipman, K.; de Alda, M.L.; et al. Future water quality monitoring — Adapting tools to deal with mixtures of pollutants in water resource management. Sci. Total Environ. 2015, 512–513, 540–551. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity concerning algae. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52].
Figure 1. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity concerning algae. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51,52].
Molecules 25 01796 g001
Figure 2. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity data concerning invertebrates. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,27,28,29,30,31,32,35,36,40,41,42,44,46,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84].
Figure 2. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity data concerning invertebrates. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,27,28,29,30,31,32,35,36,40,41,42,44,46,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84].
Molecules 25 01796 g002
Figure 3. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity data concerning fish. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,29,31,32,35,36,42,44,58,59,70,74,82,83,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137].
Figure 3. Median, maximum and minimum concentration values reported for acute (A) and chronic (B) toxicity data concerning fish. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones) [23,29,31,32,35,36,42,44,58,59,70,74,82,83,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137].
Molecules 25 01796 g003aMolecules 25 01796 g003b
Figure 4. Median and maximum risk quotients of pharmaceuticals in WWIs for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Figure 4. Median and maximum risk quotients of pharmaceuticals in WWIs for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Molecules 25 01796 g004
Figure 5. Median and maximum risk quotients of pharmaceuticals in WWEs for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Figure 5. Median and maximum risk quotients of pharmaceuticals in WWEs for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Molecules 25 01796 g005
Figure 6. Median and maximum risk quotients of pharmaceuticals in surface waters for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Figure 6. Median and maximum risk quotients of pharmaceuticals in surface waters for the three trophic levels. (Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Molecules 25 01796 g006
Figure 7. Median and maximum risk quotients of pharmaceuticals in other water bodies for the three trophic levels. (DW—drinking water; GW groundwater; SeaW—seawater; MinW—mineral water; Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Figure 7. Median and maximum risk quotients of pharmaceuticals in other water bodies for the three trophic levels. (DW—drinking water; GW groundwater; SeaW—seawater; MinW—mineral water; Anx—anxiolytics; Antib—antibiotics; Lip reg—lipid regulators; Antiepi—antiepileptics; SSRIs—Selective serotonin reuptake inhibitors; Anti-inf—anti-inflammatories; Horm—hormones).
Molecules 25 01796 g007
Table 1. Selected pharmaceuticals.
Table 1. Selected pharmaceuticals.
Therapeutic GroupCompound and Chemical Structure
Anxiolytics
(Anx)
Alprazolam (ALP)Lorazepam
(LOR)
Zolpidem
(ZOL)
Molecules 25 01796 i001 Molecules 25 01796 i002 Molecules 25 01796 i003
Antibiotics (Antib)Azithromycin (AZI)Ciprofloxacin
(CIP)
Clarithromycin (CLA)Erythromycin (ERY)
Molecules 25 01796 i004 Molecules 25 01796 i005 Molecules 25 01796 i006 Molecules 25 01796 i007
Lipid regulators
(Lip Reg)
Bezafibrate (BEZ)Gemfibrozil
(GEM)
Simvastatin
(SIM)
Molecules 25 01796 i008 Molecules 25 01796 i009 Molecules 25 01796 i010
Antiepileptic (Antiepi)Carbamazepine
(CAR)
Molecules 25 01796 i011
Selective serotonin reuptake inhibitors (SSRIs)Citalopram (CIT)Desmethylcitalopram (N-Cit) (metabolite)Escitalopram (ESC)Fluoxetine
(FLU)
Molecules 25 01796 i012 Molecules 25 01796 i013 Molecules 25 01796 i014 Molecules 25 01796 i015
Norfluoxetine (Nor-FLU) (metabolite)Paroxetine
(PAR)
Sertraline (SER)Desmethylsertraline (Nor-SER) (metabolite)
Molecules 25 01796 i016 Molecules 25 01796 i017 Molecules 25 01796 i018 Molecules 25 01796 i019
Anti-inflammatories (Anti-inf)Diclofenac
(DIC)
4-hydroxydiclofenac (4-OH-DIC) (metabolite)Ibuprofen
(IBU)
Naproxen
(NAP)
Molecules 25 01796 i020 Molecules 25 01796 i021 Molecules 25 01796 i022 Molecules 25 01796 i023
Paracetamol (PARA)4-aminophenol
(4-PARA) (transformation product)
Molecules 25 01796 i024 Molecules 25 01796 i025
Hormones (Horm)Estrone
(E1) (natural hormone/metabolite)
17β-estradiol
(E2)
17α-ethinylestradiol
(EE2)
Molecules 25 01796 i026 Molecules 25 01796 i027 Molecules 25 01796 i028
Table 2. Predicted no-effect concentrations of the selected pharmaceuticals for algae, invertebrates and fish for the studied pharmaceuticals.
Table 2. Predicted no-effect concentrations of the selected pharmaceuticals for algae, invertebrates and fish for the studied pharmaceuticals.
Therapeutic GroupPharmaceuticalPNEC (ng L−1) AlgaePNEC (ng L−1) InvertebratesPNEC (ng L−1) Fish
AnxALP892 a,b3590 b,c2540 b,c
LOR6070 a,b39,400 b,c43,100 b,c
ZOL211 a,b1550 b,c248 b,c
AntibAZI1.8 b [176] 440 e,g [83]84,000 b
CLA2 b [23]8160 b [23]1,000,000 b [23]
CIP5 b [35]10,000 b [44]1,000,000 b [44]
ERY20 b [23]220 b [23]1,000,000 b [23]
Lip regBEZ4870 a,b1300 e,f [51]17,600 b,c
GEM15,190 b [51]1180 b [53]150 e,g [85]
SIM22,800 b [26]3.2 d [56]765 b,c
AntiepiCAR31.6 b [27]0.2 d [57]20,000 b [44]
SSRIsCIT1600 b [30]3900 b [58]4470 b,c
FLU44.99 b [33]2 d [57]2.8 e,g [177]
Nor-FLU (M)189 b [34]300 b [61]n.a
PAR140 b [30]580 b [58]3290 b,c
SER12.10 b [33]120 b [58]72 b [92]
Anti-infDIC200 d [37]20,000 e,g [70]50 e,g [95]
4-OH-DIC (M)660,300 e,f [38]48,200 b,c65,200 b,c
IBU40,100 e,f [39]0.2 d [57]180 e,g
NAP31,820 b [40]2620 b [53]115,200 b [99]
PARA134,000 b [59]2040 b [73]378,000 b [42]
4-PARA (TP)11,300 a,b240 b [77]1430 b [100]
HormE1 (NH/M)355 a,b3160 b,c3.4 e,g [103]
E2162 a,b1500 b [78]0.29 e,g [113]
EE2730 b [43]10 e,g [81,82]0.01 e,g [123]
M—metabolite; TP—transformation product; NH—natural hormone. a: EC50 was estimated with ECOSAR. b: UF = 1000. c: LC50 was estimated with ECOSAR. d: UF = 50 (uncertainty factor used for lowest observed effect concentrations (LOEC) and no observed effect concentrations (NOEC) in acute toxicity). e: long-term data. f: UF = 100. g: UF = 10 (uncertainty factor used for LOEC and NOEC in chronic toxicity).

Share and Cite

MDPI and ACS Style

Pereira, A.; Silva, L.; Laranjeiro, C.; Lino, C.; Pena, A. Selected Pharmaceuticals in Different Aquatic Compartments: Part II—Toxicity and Environmental Risk Assessment. Molecules 2020, 25, 1796. https://doi.org/10.3390/molecules25081796

AMA Style

Pereira A, Silva L, Laranjeiro C, Lino C, Pena A. Selected Pharmaceuticals in Different Aquatic Compartments: Part II—Toxicity and Environmental Risk Assessment. Molecules. 2020; 25(8):1796. https://doi.org/10.3390/molecules25081796

Chicago/Turabian Style

Pereira, André, Liliana Silva, Célia Laranjeiro, Celeste Lino, and Angelina Pena. 2020. "Selected Pharmaceuticals in Different Aquatic Compartments: Part II—Toxicity and Environmental Risk Assessment" Molecules 25, no. 8: 1796. https://doi.org/10.3390/molecules25081796

Article Metrics

Back to TopTop